The Pan Histone Deacetylase Inhibitor Panobinostat and the Iso-Selective Inhibitor Romidepsin Reduce Glycoprotein VI Expression and Inhibit Platelet Activation in Response to Collagen Related Peptide

Blood ◽  
2011 ◽  
Vol 118 (21) ◽  
pp. 1154-1154
Author(s):  
Mark J Bishton ◽  
Miles Prince ◽  
Ricky W Johnstone ◽  
Simon J. Harrison ◽  
Benjamin T. Kile

Abstract Abstract 1154 Histone deacetylase inhibitors (HDACi) are anti-cancer drugs able to induce chromatin remodelling, alter gene expression and affect function of non-histone proteins. We recently reported that the pan-HDACi panobinostat and the iso-selective inhibitor romidepsin induce thrombocytopenia by reducing megakaryocyte proplatelet number, without effects on platelet half life (Bishton et al Blood 2011). The effect of HDACis on platelet function remains unknown, and we postulated possible interference with the expression or function of key platelet activating proteins. Platelet Glycoprotein (GP) VI is a member of the immunoglobulin superfamily, expressed exclusively on the surface of platelets and megakaryocytes, complexed with FcR g-chain dimers, predominantly responsible for adhesion of platelets to collagen. Following interaction with sub-endothelial collagen, the Src family kinases Fyn and Lyn mediate the recruitment and autophosphorylation of Syk kinase and thereby downstream signalling and platelet activation. Following the treatment of C57BL/6 mice with 10mg/kg panobinostat or 1mg/kg romidepsin intraperitoneally (IP) daily for three days, we isolated washed murine platelets for function testing. Following stimulation with thrombin, a dose-dependant increase was seen in platelet surface expression of CD62P (P-selectin), and also the conformationally active form of the integrin αIIbbIIIa, with no difference seen between groups. When collagen related peptide (CRP) was used as a platelet agonist, and activation assessed by p-selectin and activated αIIbbIIIa expression, platelets from cohorts of mice treated with panobinostat or romidepsin failed to increase the expression of either molecule in response to CRP, compared to vehicle treated mice. Co-treatment of mice with the murine thrombopoietin mimetic, AMP-4, or the proteasome inhibitor bortezomib did not alter effects of the HDACi. Ex vivo addition of panobinostat or romidepsin to naïve platelets did not however affect platelet activation, suggesting megakaryocytes rather than platelets to be the target cell responsible for these effects. Flow cytometric analysis of the expression of GPVI on platelets showed a consistent and statistically significant decrease in the median fluorescent intensity (MFI) of staining seen in both HDACi treated groups. No equivalent changes in the surface expression of the other collagen receptor integrin α2b1 were seen. Western blotting of murine platelets confirmed this reduction in GPVI and a ∼17kDa fragment was also seen with HDACi treated platelets, suggesting GPVI degradation. Following stimulation with CRP, Western blotting of platelets with a phospho-syk antibody showed a reduction in phospho-syk levels in platelets from mice treated with HDACi, consistent with decreased downstream signalling from the GPVI receptor. Western blotting of murine megakaryocytes differentiated from murine fetal liver cells by TPO, also demonstrated a reduction in GPVI expression following HDACi exposure, again suggesting an intrinsic megakaryocyte process to be responsible. qRT-PCR on HDACi treated megakaryocytes demonstrated a mild increase in GPVI mRNA levels post romidepsin, but no changes post panobinostat compared to vehicle treated cells, confirming transcriptional repression not to be responsible for these changes. We show that HDACi cause a reduction in surface expression of GPVI expression by inducing its degradation and thus inhibiting murine platelet responses to CRP. There was no evidence of an effect on gene transcription. Our work suggests a potential beneficial anti-thrombotic effect of HDACi, mediated by reduction in both platelet number and function. These findings support the need to investigate the role of HDACi and their effect on GPVI in myeloproliferative neoplasms particularly with respect to their impact on thrombotic complications. Disclosures: Off Label Use: Panobinostat and romidepsin are histone deacetylase inhibitors. We show that both reduce platelet response to collagen and therefore may have an anti-thrombotic effect. Prince:Novartis: Membership on an entity's Board of Directors or advisory committees, Research Funding; Cellgene: Membership on an entity's Board of Directors or advisory committees, Research Funding. Johnstone:N: Research Funding. Harrison:Cellgene: Honoraria, Membership on an entity's Board of Directors or advisory committees, Research Funding; Novartis: Research Funding.

Blood ◽  
2011 ◽  
Vol 118 (21) ◽  
pp. 811-811 ◽  
Author(s):  
Meletios Athanasios Dimopoulos ◽  
Sundar Jagannath ◽  
Sung-Soo Yoon ◽  
David S. Siegel ◽  
Sagar Lonial ◽  
...  

Abstract Abstract 811 Introduction: Vorinostat (VOR), an oral inhibitor of histone deacetylase class I and class II proteins, regulates genes and proteins involved in tumor growth and survival. The synergistic effects of VOR and bortezomib (BTZ) have been shown in preclinical studies and were confirmed in independent phase 1 trials in patients with relapsed/refractory multiple myeloma (MM), producing objective response rates (ORRs) of up to 42% and overall clinical benefit of up to 90%. Materials and methods: Eligible patients were aged ≥ 18 years, had measurable secretory MM, had received 1 to 3 prior systemic anti-myeloma regimens, and had an Eastern Cooperative Oncology Group status ≤ 2. Previous exposure to BTZ and the presence of extracellular plasmacytoma were allowed per protocol, but patients with prior resistance to BTZ were excluded. Patients were randomized 1:1 to receive 21-day cycles of BTZ (1.3 mg/m2 intravenously; days 1, 4, 8, and 11) in combination with oral VOR 400 mg/d, or matching placebo, on days 1 to 14. Additional use of corticosteroids for the treatment of MM was not allowed during the trial. Patients were treated until disease progression, unacceptable toxicities, or withdrawal from the study. The primary endpoint for this trial was progression-free survival (PFS; occurrence of 412 PFS events). Secondary and exploratory endpoints included ORR (≥ partial response), clinical benefit response (ORR + minimal response), overall survival, time to progression, patient-reported outcomes questionnaires (QLQ-C30, QLQ-MY20), and safety/tolerability of this novel drug combination. Responses and progression were determined according to the European Bone and Marrow Transplantation Group criteria and will be confirmed by an Independent Adjudication Committee. Results: Between January 2009 and January 2011, 637 patients were enrolled from 174 centers in 33 countries across the globe making this trial one of the largest studies conducted in patients with relapsed/refractory myeloma. Median age of the study population was 62 years (range, 29–86 years). Of the enrolled patients, 59% were male and 56% were Caucasian. Patients had received a median of 2 prior regimens (range, 1–3). Prior anti-myeloma agents included BTZ (24%), thalidomide (56%), lenalidomide (13%), melphalan (56%), and stem cell transplantation (35%). As of July 2011, 635 patients had received study medication, with a median exposure of 7 cycles (mean: 7.6 cycles; range 1–30 cycles). Reported median exposure to BTZ monotherapy in previous phase 3 trials was approximately 5 cycles. Conclusions: The study passed the protocol-specified futility analyses by the independent data monitoring committee in November 2010. Database lock is anticipated in November 2011, and top-line data on primary and secondary endpoints will be available at the meeting. Disclosures: Dimopoulos: Celgene, Ortho-Biotech: Consultancy, Honoraria. Off Label Use: Vorinostat, an inhibitor of histone deacetylase, is approved in the US for the treatment of cutaneous manifestations in patients with cutaneous T cell lymphoma (CTCL) who have progressive, persistent or recurrent disease on or following two systemic therapies. Vorinostat is currently under investigation for the treatment of relapsed malignant pleural mesothelioma, relapsed/refractory B cell lymphoma (in combination with other chemotherapy agents), and relapsed/refractory multiple myeloma (in combination with bortezomib and other chemotherapy agents). Jagannath:Merck: Membership on an entity's Board of Directors or advisory committees; Celgene: Membership on an entity's Board of Directors or advisory committees; Millennium: Membership on an entity's Board of Directors or advisory committees. Yoon:Celgene: Consultancy; NK Bio: Consultancy. Siegel:Millennium: Honoraria, Research Funding, Speakers Bureau; Merck: Honoraria. Lonial:Millennium: Consultancy; Celgene: Consultancy; Novartis: Consultancy; BMS: Consultancy; Onyx: Consultancy; Merck: Consultancy. Hajek:Celgene: Honoraria; Janssen: Honoraria; Merck: Educational lecture. Facon:Janssen: Membership on an entity's Board of Directors or advisory committees, Speakers Bureau; Merck: Membership on an entity's Board of Directors or advisory committees. Rosiñol:Celgene: Honoraria; Janssen-Cilag: Honoraria. Blacklock:New Zealand Bone Marrow Donor Registry: Consultancy, Employment; Mercy Hospital, Auckland New Zealand: Consultancy; Leukaemia and Blood Foundation, New Zealand: Consultancy, Membership on an entity's Board of Directors or advisory committees; Middlemore Hospital: Employment, Research Funding. Goldschmidt:Amgen, Novartis, Chugai: Research Funding; Janssen-Cilag, Celgene: Membership on an entity's Board of Directors or advisory committees, Research Funding. Palumbo:Merck: Honoraria; Celgene: Honoraria, Membership on an entity's Board of Directors or advisory committees; Amgen: Honoraria; Janssen-Cilag: Honoraria, Membership on an entity's Board of Directors or advisory committees. Reece:Merck: Honoraria, Research Funding; Janssen: Honoraria, Research Funding; Celgene: Honoraria, Research Funding; Otsuka: Honoraria, Research Funding. Graef:Merck: Employment. Houp:Merck Research Laboratories: Employment. Sun:Merck & Co., Inc.: Employment. Eid:Merck Research Laboratories: Employment. Anderson:Celgene: Consultancy; Millennium: Consultancy; Novartis: Consultancy; BMS: Consultancy; Onyx: Consultancy; Merck: Consultancy; Acetylon: founder.


Blood ◽  
2013 ◽  
Vol 122 (21) ◽  
pp. 2823-2823 ◽  
Author(s):  
Catriona HM Jamieson ◽  
Robert P Hasserjian ◽  
Jason Gotlib ◽  
Jorge E. Cortes ◽  
Richard M. Stone ◽  
...  

Abstract Introduction Fedratinib, a JAK2-selective inhibitor, demonstrated clinical benefit through a reduction in splenomegaly and symptoms in patients with myelofibrosis (MF), including post-polycythemia vera MF (post-PV MF), post-essential thrombocythemia MF (post-ET MF) and primary MF (PMF), in Phase I and II studies (J Clin Oncol 2011;29:789; Haematologica 2013;98:S1113). Bone marrow fibrosis (BMF) has been associated with splenomegaly and cytopenias (Ann Hematol 2006;85:226). Hence, stabilization and/or reversal of BMF remain important therapeutic goals. This report represents an exploratory analysis of sequential BMF data from patients with MF in an open-label Phase I/II study to evaluate the long-term effects of orally administered fedratinib (TED12015; NCT00724334). Methods Patients with intermediate or high-risk MF (Mayo Prognostic Scoring System) received fedratinib therapy in consecutive cycles (1 cycle = 28 days) as long as they derived clinical benefit. Bone marrow trephine biopsies were performed at baseline and after every 6 cycles. Hematoxylin and eosin, reticulin, and Masson's trichrome staining of core biopsy slides were used to grade BMF on a scale from 0 to 3 using the 2008 WHO MF grading criteria. BMF was graded independently in a blinded fashion by 3 hematopathologists. BMF grades were established as long as at least 2 of the 3 pathologists agreed independently. Changes in BMF grade from baseline were categorized as improvement (≥1 grade reduction), stabilization (no change), or worsening (≥1 grade increase). Results Of the 43 patients enrolled in the TED12015 study, the median fedratinib dose received was 473 (range 144–683) mg/day and median treatment duration was 32.3 (range 7–61) cycles. Bone marrow biopsies at baseline and at least one other time point were available for 21/43 (49%) patients, whose baseline characteristics were: median age 61 years (range 43–85); 57% male; 38% high-risk MF by WHO 2008 criteria (Leukemia 2008; 22:14); and 90% JAK2V617F positive. A consensus grade was achieved for 96% of the samples. At baseline, 2, 10, and 9 patients had grade 1, 2, and 3 BMF, respectively. Changes in BMF grade from baseline are shown in the figure. BMF improvement with 1 grade reduction was observed in 8/18 (44%) patients at Cycle 6. By Cycle 30, 4/9 (44%) evaluable patients had BMF improvement, including 2 patients with improvement by 2 grades and 2 patients with improvement by 1 grade. Of patients with Grade 3 BMF at baseline, 6/9 (67%) exhibited 1 grade improvement at Cycle 6. Two patients had 2 grades of BMF reduction from baseline during treatment (grade 3 to 1, and grade 2 to 0, both at Cycle 12), and the latter achieved a complete clinical remission at Cycle 30 assessed by IWG-MRT response criteria. The two patients who experienced complete reversal of BMF to grade 0 (one from grade 2 and one from grade 1) had normalization of not only hemoglobin level but also white blood cell and platelet counts at Cycle 18. Conclusions These exploratory analyses suggest that a proportion of patients treated long-term with fedratinib demonstrate stable or improved BMF. The disease modifying impact of fedratinib on BMF changes will be further assessed in a randomized, placebo-controlled Phase III clinical trial (JAKARTA; NCT01437787). This study was sponsored by Sanofi. Disclosures: Jamieson: J&J, Roche: Research Funding; Sanofi: Membership on an entity’s Board of Directors or advisory committees. Hasserjian:Sanofi, Inc: Consultancy. Gotlib:Sanofi: Travel to EHA 2012, Travel to EHA 2012 Other; Sanofi: Membership on an entity’s Board of Directors or advisory committees; Sanofi: Research Funding. Cortes:Incyte, Sanofi: Consultancy; Incyte, Sanofi: Research Funding. Talpaz:Novartis, Bristol-Myers Squibb, Ariad, Deciphera: Research Funding; Novartis, Bristol-Myers Squibb, Ariad, Deciphera: Speakers Bureau. Thiele:AOP Orphan Pharmaceuticals, Incyte, Novartis, Shire, Sanofi: Consultancy; Novartis, Shire: Research Funding; AOP Orphan Pharmaceuticals, Incyte, Novartis, Shire, Sanofi: Honoraria. Rodig:Ventana/Roche Inc.: Research Funding; Daiichi-Sankyo/Arqule Inc., Ventana/Roche Inc., Shape Pharmaceuticals Inc.: Consultancy. Patki:Sanofi: Employment. Wu:Sanofi: Employment. Wu:Sanofi: Employment. Pozdnyakova:Sanofi: Honoraria; Sanofi: Consultancy.


Blood ◽  
2016 ◽  
Vol 128 (22) ◽  
pp. 3949-3949 ◽  
Author(s):  
Melissa A. Fischer ◽  
Sharon Friedlander ◽  
Leah Hogdal ◽  
Pia Arrate ◽  
Hua Chang ◽  
...  

Abstract Introduction: Exportin 1 (XPO1) is a well characterized and essential nucleo-cytoplasmic transport protein in the karyopherin family, and is responsible for the nuclear export of over 200 cargo proteins, including the major tumor suppressor proteins (TSPs) p53, p21, FOXO and the translation regulator elF4E. XPO1 is overexpressed in numerous cancer types including solid and hematological malignancies, often correlating with poor prognosis. Recently, a novel class of Selective Inhibitors of Nuclear Export (SINE) compounds, selinexor (KPT-330) and the second generation KPT-8602, have been developed for the treatment of advanced cancers. We have previously shown that selinexor has marked activity in AML and DLBCL pre-clinical models. The BCL-2 family of anti-apoptotic proteins are deregulated and linked to maintenance and survival in AML and DLBCL. For its translation, the mRNA for BCL-2 is transported from the nucleus to the cytoplasm by forming a complex with XPO1 cargo, elF4E. Other important mRNAs exported from the nucleus via this mechanism include BCL6 and MYC. We hypothesize that SINE compounds inhibit XPO1/elF4E-mediated nuclear-cytoplasmic transport by covalently binding to the XPO1 cargo binding site and that in the absence of protein translation, BCL-2, BCL6 and MYC levels rapidly decline. Venetoclax (VEN; ABT-199) is a potent, selective inhibitor of BCL-2. In vitro, AML cells acquire resistance to VEN over time, often due to up-regulation of another BCL-2 family anti-apoptotic protein, MCL-1. MCL-1 is regulated by the anti-apoptotic transcription factor and XPO1 cargo NF-kB. We have previously shown that SINE compounds significantly decreased MCL-1 levels, presumably via inactivation of NF-kB. The goal of this study was to test whether SINE compounds will synergize with VEN via BCL-2 modulation and whether the combination would diminish MCL-1 mediated resistance to BCL-2 inhibition in DLBCL and AML models, respectively. Methods: BH3 profiling was performed in a sample of cell lines using a cytochrome c release assay to identify anti-apoptotic dependencies. The effects of SINE compounds and VEN as single agents or in combination on cell viability were performed in AML (K-562, MOLM-13, MV-4-11, and U-937) and DLBCL cell lines (SU-DHL-6, DoHH-2 and Toledo). Whole cell protein lysates were extracted 24 hours after treatment for immunoblot analysis. The activity of SINE compounds (5 mg/kg) and VEN (25 mg/kg) as single agents, or in combination were measured in AML (MV-4-11) and DLBCL (DoHH-2 and Toledo) xenografts in NSGS and nude mice, respectively. Tumor growth and survival were measured throughout these animal studies. Tumor tissue was collected at the end of treatment for flow cytometric analysis, western blotting and immunohistochemistry (IHC). Results: By employing BH3 profiling, we identified AML cell lines that were dependent (MV-4-11 and MOLM-13) and not dependent (U-937 and K-562) on MCL-1. Dose response analysis demonstrated that each of the AML cell lines was sensitive to the SINE compounds, while VEN only reduced viability in the MV-4-11 and MOLM-13 cells. Additionally, there was enhanced growth inhibition when the SINE compounds were combined with VEN in the MCL-1 dependent cells. SINE compound treatment synergistically decreased c-MYC protein levels in all 4 AML cell lines with the combination treatment (Figure 1), whereas PARP cleavage was only enhanced with the combination in the MV-4-11 and MOLM-13 cells. Likewise, MCL-1 is reduced in the presence of SINE compound or SINE compound-VEN combinations. In DLBCL xenograft studies (DoHH-2 and Toledo), combination of selinexor with VEN was synergistic for tumor reduction and increased animal survival when compared to either single agent alone. By IHC we observed a concomitant reduction in BCL-2 and BCL-6 and an increase in cleaved caspase 3 in DLBCL tumors after combination treatment. Conclusions: SINE compound-VEN combinations show enhanced antitumor effect, with reduction of oncogenic activity. SINE compounds reduce MCL-1 in VEN-resistant cells. As MCL-1 driven anti-apoptotic machinery is responsible for resistance to inhibition of BCL-2 in DLBCL and AML, SINE compound regulation of MCL-1 may lead to rescue of VEN resistance. SINE compounds and VEN are excellent candidate partners for combination therapies in AML and DLBCL. Disclosures Friedlander: Karyopharm Therapeutics: Employment. Chang:Karyopharm Therapeutics: Employment, Equity Ownership. Kashyap:Karyopharm Therapeutics: Employment, Equity Ownership. Argueta:Karyopharm Therapeutics: Employment, Equity Ownership. Klebanov:Karyopharm Therapeutics: Employment, Equity Ownership. Senapedis:Karyopharm Therapeutics: Employment, Equity Ownership. Baloglu:Karyopharm Therapeutics: Employment, Equity Ownership. Lee:Karyopharm Therapeutics: Employment, Equity Ownership. Shacham:Karyopharm Therapeutics: Employment, Equity Ownership, Membership on an entity's Board of Directors or advisory committees. Savona:TG Therapeutics: Research Funding; Amgen Inc.: Membership on an entity's Board of Directors or advisory committees; Takeda: Research Funding; Sunesis: Research Funding; Incyte: Membership on an entity's Board of Directors or advisory committees, Research Funding; Celgene: Membership on an entity's Board of Directors or advisory committees; Ariad: Membership on an entity's Board of Directors or advisory committees; Gilead: Membership on an entity's Board of Directors or advisory committees.


Blood ◽  
2021 ◽  
Vol 138 (Supplement 1) ◽  
pp. 2539-2539
Author(s):  
Kyle H Vining ◽  
Anna E. Marneth ◽  
Kwasi Adu-Berchie ◽  
Christina M. Tringides ◽  
Joshua M Grolman ◽  
...  

Abstract Myelofibrosis (MF) is a progressive, myeloid malignancy characterized by deposition of collagen and reticulin fibers in the bone marrow (BM). Previous studies have shown that monocytosis is associated with poor prognosis in MF, highlighting a potential pathogenic role for monocytes in MF. Although many studies have addressed the role of cell-intrinsic and soluble extracellular factors in MF development, it is currently unknown if mechanical properties of fibrotic BM contribute to aberrant differentiation of myeloid cells and of monocytes in particular. We first defined the stiffness and viscoelastic properties of healthy and fibrotic BM. Stiffness is defined as the resistance of a matrix to deformation, while viscoelasticity is the rate of dissipation of an applied stress over time. Independent of stiffness, an applied stress relaxes rapidly in a more viscous, liquid-like matrix, whereas in a more elastic, solid-like material, stress relaxes slowly. We next generated a cohort of fibrotic and non-fibrotic mice by transplanting retrovirally transduced JAK2V617F or empty vector (EV) control hematopoietic stem and progenitor cells (HSPCs) into lethally irradiated recipients. Femurs from these mice were harvested seven months post-transplant, as well as from age- and sex-matched healthy primary mice. Nanoindentation was performed to measure BM stiffness and viscoelasticity. Fibrotic BM showed higher stiffness, as well as trending higher elastic, solid-like properties, compared to BM of control mice. We then aimed to study the effect of matrix stiffness and viscoelasticity on monocytes. Human BM-derived monocytes were encapsulated in stiff, viscous or stiff, elastic hydrogels and cultured in the presence of GM-CSF, IL-4, and PGE2 for 3 days, followed by nanoString and flow cytometry analyses. Cells in elastic gels upregulated gene sets associated with co-stimulatory molecules and cytokine receptor signaling, MHC class II antigen presentation, and regulation of extracellular matrix (ECM), compared to cells in viscous gels of the same stiffness. The fraction of dendritic cells (DCs) was significantly upregulated, as indicated by double-positive CD11c+CD1c+ (40.9% viscous vs 69.5% elastic of CD11b+HLA-DR+ cells) and CD80+ cells (20.9% viscous vs 62.7% elastic of CD11b+HLA-DR+ cells), and surface expression of HLA-DR (gMFI 2587 viscous vs 6334 elastic). Consistent with these findings, the fraction of pro-fibrotic SLAMF7+ cells (4.2% viscous vs 17.3% elastic) were also significantly higher in elastic gels. Together, these data suggest that stiff, elastic ECM drives pro-inflammatory polarization and differentiation of monocytes into antigen-presenting cells. Next, we examined the role of the cytoskeleton on human monocyte differentiation. Cortical F-actin was significantly upregulated in cells in stiff, elastic gels compared to viscous gels. Cells were exposed to a highly selective small molecular inhibitor of the γ-isoform of PI3K. Treatment with the PI3Ky inhibitor significantly reduced F-actin staining of cells in elastic gels, upregulated immature monocyte markers, reduced surface expression of HLA-DR, and downregulated the cytokines IL6, IL8, CCL4, which have previously been associated with disease progression in myelofibrosis. In line with the above human ex vivo data, BM isolated from fibrotic mice (described above) showed skewing towards Ly6G-Ly6C+ monocytes (a population enriched for inflammatory monocytes) within the CD11b myeloid compartment compared to control transplanted mice or to non-fibrotic mice that were transplanted with endogenously expressing Jak2V617F cells. Additionally, the percentage of conventional DCs (cDCs) was increased in fibrotic Jak2V617F mice compared to control mice. Importantly, 16 day in vivo treatment with the PI3Ky inhibitor significantly reduced the fraction of Ly6G-Ly6C+ monocytes within the CD11b compartment as well as the fraction of cDCs, compared to vehicle-treated Jak2V617F mice. In summary, fibrotic BM is stiffer and more elastic than normal BM. Our studies show that a stiff, elastic BM environment drives monocytes towards a more pro-inflammatory state which can in part be suppressed by PI3K-γ inhibition. Our results have relevance for human MF by demonstrating that a fibrotic BM niche is not just a consequence of chronic inflammation but is also inflammation-promoting. KHV and AEM contributed equally to this work. Disclosures Pozdnyakova: Scopio Labs: Consultancy. Mullally: Janssen, PharmaEssentia, Constellation and Relay Therapeutics: Consultancy. Wucherpfennig: Novartis: Research Funding; Nextechinvest: Membership on an entity's Board of Directors or advisory committees; Immunitas Therapeutics: Current holder of individual stocks in a privately-held company; TScan Therapeutics: Membership on an entity's Board of Directors or advisory committees; TCR2 Therapeutics: Membership on an entity's Board of Directors or advisory committees; SQZ Biotech: Membership on an entity's Board of Directors or advisory committees. Mooney: Novartis: Patents & Royalties: Licensed IP, Research Funding; Sirenex: Patents & Royalties: Licensed IP; Samyang Corp: Membership on an entity's Board of Directors or advisory committees; IVIVA: Membership on an entity's Board of Directors or advisory committees; Attivare: Current holder of individual stocks in a privately-held company, Membership on an entity's Board of Directors or advisory committees; Revela: Membership on an entity's Board of Directors or advisory committees; Amend Surgical: Patents & Royalties: Licensed IP; Lyell: Current equity holder in publicly-traded company, Patents & Royalties.


Blood ◽  
2019 ◽  
Vol 134 (Supplement_1) ◽  
pp. 4203-4203
Author(s):  
Nicole Kucine ◽  
Amanda R. Leonti ◽  
Aishwarya Krishnan ◽  
Rhonda E. Ries ◽  
Ross L. Levine ◽  
...  

Introduction : Myeloproliferative neoplasms (MPNs) are rare clonal bone marrow disorders in children characterized by high blood counts, predisposition to clotting events, and the potential to transform to myelofibrosis or acute myeloid leukemia (AML). Children with MPNs have lower rates of the known driver mutations (in JAK2, MPL, and CALR) than adult patients, and the underlying pathways and molecular derangements in young patients remain unknown. Given the lack of knowledge about pediatric MPNs, it is critical that we gain a better understanding of the dysregulated pathways in these diseases, which is necessary for improving disease understanding and broadening treatment options in children. Therefore, the objective of this work was to identify differentially expressed genes and pathways between children with MPNs and healthy controls, as well as children with AML, to guide further study. Methods : Mononuclear cells were extracted from peripheral blood of pediatric MPN patients (n=20) and pediatric and young adult AML patients (n=1410), and bone marrow of normal controls (NC, n=68). AML patient samples were being evaluated as part of a Children's Oncology Group planned analysis. To identify an expression profile unique to MPNs, transcriptome data from MPN patients was contrasted against NC and AML patients. All samples were ribodepleted and underwent Illumina RNA-Seq to generate transcriptome expression data. All analyses were performed in R. Differentially expressed genes were identified using the voom function from the limma package (v. 3.38.3), and enriched pathways were identified using the pathfindR package (v. 1.3.1). Unsupervised hierarchical clustering and heatmap generation was performed using the ComplexHeatmap package (v. 1.20.0). Results : MPN patient samples showed a unique expression signature, distinct from both AML patients and normal controls. Unsupervised PCA plot (Figure 1A) and heatmaps (Figure 1B) show that MPN samples cluster together. There were 4,012 differentially expressed (DE) genes in MPNs compared to NC and 6,743 DE genes in MPNs compared to AML patients. There were 2,493 shared genes between the 2 groups (Figure 1C.) Significantly DE genes between MPNs and other groups included multiple platelet-relevant genes including PF4 (CXCL4), PF4V1, P2RY12, and PPBP (CXCL7). Interestingly, PF4V1 was the most DE gene in MPNs compared to AML, and third highest versus NC. Dysregulation of some of these genes has been seen in adult MPNs, as well as thrombosis. Further comparison of transcriptome profiles between children with (n=13) and without (n=7)JAK2 mutations showed upregulation of three genes, CFB, C2, and SERPING1, which are all known complement genes, implicating complement activation in JAK2-mutated MPN patients. Complement activation has previously been reported in adult MPNs. Pathway enrichment analysis shows a number of immune and inflammatory pathways as enriched in MPN patients compared to both AML and NC. There were 179 enriched pathways in MPNs compared to AML and 142 compared to NC, with 134 common pathways (Figure 1D.) The systemic lupus erythematosus pathway was the most heavily enriched pathway in MPNs compared to both AML and NC. Additional pathways with significant enrichment include hematopoietic cell lineage, cytokine-cytokine interactions, DNA replication, and various infection-relevant pathways. The JAK-STAT signaling pathway was also enriched in MPNs compared to both AML and NC, as was the platelet activation pathway. Conclusion: Transcriptome evaluation of childhood MPNs shows enrichment of numerous inflammatory and immune pathways, highlighting that, as in adult MPNs, inflammation is implicated in pediatric MPNs. Furthermore, specific complement genes were upregulated in JAK2-mutant MPN. Upregulation of platelet-specific genes implies potential insights into disease mechanisms and warrants more study. Variations in the cell populations may account for some of the differences seen, however all samples were largely mononuclear cells, making their comparisons reasonable. Further analysis of this early data is needed to better assess inflammatory changes and platelet activation in pediatric MPNs, as are larger sample sizes. Individual cells may have differential expression of various genes, and future experiments with single-cell RNA-seq would be helpful to further elucidate differences. Disclosures Levine: Novartis: Consultancy; Loxo: Membership on an entity's Board of Directors or advisory committees; Celgene: Consultancy, Research Funding; Gilead: Consultancy; Roche: Consultancy, Research Funding; Lilly: Honoraria; Amgen: Honoraria; Qiagen: Membership on an entity's Board of Directors or advisory committees; Imago Biosciences: Membership on an entity's Board of Directors or advisory committees; C4 Therapeutics: Membership on an entity's Board of Directors or advisory committees; Prelude Therapeutics: Research Funding; Isoplexis: Membership on an entity's Board of Directors or advisory committees.


Blood ◽  
2010 ◽  
Vol 116 (21) ◽  
pp. 5037-5037
Author(s):  
Sam A Ruell ◽  
Miles Prince ◽  
Hang Quach ◽  
Emma Link ◽  
Joanne Dean ◽  
...  

Abstract Abstract 5037 Various bortezomib (Bz)-based combinations are being examined in the context of relapsed and/ or refractory Multiple Myeloma (MM). Histone Deacetylase Inhibitors (HDACIs) are a novel group of agents that effect a variety of cellular and intracellular processes by enhancing the acetylation of histone and non-histone targets. Romidepsin (Romi), a cyclic tetrapeptide, is a class 1 HDACi. Initial reports suggested that various HDACi induced QTc prolongation as a class effect, although subsequently it has been shown that careful electrolyte management prevent this from being a clinical problem. 32 patients (pts) have been enrolled in a Phase I/II Trial of Romi, Bz and dexamethasone (Dex) in relapsed or refractory MM. The maximum tolerated dose (Romi 10mg/m2, Bz 1.3mg/m2 and Dex 20mg) was determined in the first 6 pts with Romi given on day 1, 8 & 15, Bz on D1, 4, 8 and 11 and Dex D1, 2, 4, 5, 8, 9, 11 and 12 of a 28 day cycle (n=25). In an ongoing expanded Phase IIb cohort Romi is only given on day 1 & 8 of a 21 day cycle (n=7/15). Romidepsin was initially given as the standard 4 hour infusion. However animal and clinical safety data support the use of 1hr Romi infusions. In the expansion cohort, the duration of Romi infusions was reduced to 1 hour from cycle 2. To monitor safety (specifically QT interval), ECGs were mandatory pre and post the 4 hour infusions in cycle one and the 1 hour infusions in cycle 2 and reviewed prior to the continuation of the 1 hour infusion from cycle 3. Prolongation of QTc was defined as an increase of 33%, or 60 msec, or QTc 3500 msec. The maintenance of serum potassium > 4.0 mmol/L and magnesium >0.85 mmol/L prior to Romi infusion was mandated. Any patients in the initial cohort remaining on study and still receiving Romi were eligible to move to 1 hour Romi infusions with ECG monitoring during the first cycle. 32 patients have received 598 Romi infusions. 525 infusions have been given over 4 hours and 73 over 1 hour. 19 pts only received 4 hour infusions and 13 pts received at least one 1 hour infusion of Romi. The phase I/II cohort (n=25, median lines of therapy 2 (range1-3)) received 4 hour infusions, the overall response (CR+PR+MR) by modified EBMT criteria was 76% in 21 assessable patients. 2pts had CR (10%), 13 PR (62%), 1 MR (4%), 4 pts had SD, and 1PD. The Phase IIb cohort (n=7, median lines of therapy 2 (range 1–4)) received 1 hour infusions and 6 pts are currently assessable for response with 3 PR and 3 MR. In the first 25 pts receiving the 4 hour infusions, 2 patients experienced arrhythmias during their first cycle, grade 2 atrial flutter possibly related to drug and sinus tachycardia definitely not related. 1pt developed severe ischaemic heart disease and 1pt has died, likely from a PE. Since the introduction of 1hr Romi infusions, only one patient has experienced a Grade 1 cardiac event of asymptomatic lateral T wave flattening and mild ST depression on ECG. This occurred in cycle one, with 4 hour infusions. Full cardiac assessment reveled no clinically significant abnormality. This patient went on to receive the 1 hour infusions without incident. The implementation of the 1 hour infusions has successfully reduced the overall treatment time with Romi/ Bz/ Dex without increase in toxicity. Although numbers are small the response rates appear similar to those using the 4 hour infusions. Disclosures: Off Label Use: Romidepsin and Velcade in combination for the treatment of relapsed/refractory myeloma. Prince:Celgene: Honoraria, Membership on an entity's Board of Directors or advisory committees, Research Funding. Harrison:Celgene: Honoraria, Membership on an entity's Board of Directors or advisory committees, Research Funding.


Blood ◽  
2011 ◽  
Vol 118 (21) ◽  
pp. 480-480 ◽  
Author(s):  
David S. Siegel ◽  
Meletios Athanasios Dimopoulos ◽  
Sung-Soo Yoon ◽  
Jacob P. Laubach ◽  
Jonathan L. Kaufman ◽  
...  

Abstract Abstract 480 Introduction: Vorinostat (VOR), an oral inhibitor of histone deacetylase (HDAC) class I and class II proteins, affects pathways regulating cell proliferation and apoptosis in a diverse array of tumor types. Phase 1/2 trials in multiple myeloma (MM) have indicated clinical activity of vorinostat, as both a single agent and in combination with bortezomib (BTZ) and immunomodulatory drugs (IMiDs). Materials and methods: The present study (MK-0683 PN095) was an open-label, single-arm phase 2b trial of VOR plus BTZ in BTZ-refractory patients (defined as < 25% response on therapy, or progression during or < 60 days after completion of therapy) and patients considered to be refractory, intolerant, or ineligible for IMiD-based therapy regimens. Eligible patients were aged ≥ 18 years, had measurable secretory MM, had received ≥ 2 prior anti-myeloma regimens, and relapsed or progressed following prior systemic therapy. Patients received 21-day cycles of BTZ (1.3 mg/m2 intravenously [IV]; days 1, 4, 8, and 11) plus oral VOR 400 mg/d on days 1 to 14. If patient had no change as the best response after 4 cycles of treatment or progressive disease after 2 cycles of treatment, oral dexamethasone 20 mg on the day of and day after each dose of BTZ could be added to the treatment regimen. Patients were treated until disease progression, unacceptable toxicities, or withdrawal from the study. The primary endpoint was overall response rate (ORR; ≥ partial response). Secondary and exploratory endpoints included clinical benefit response (ORR + minimal response), overall survival (OS), time to progression (TTP), progression-free survival (PFS), and safety. Responses and progression were determined according to the International Myeloma Working Group criteria and the European Bone and Marrow Transplantation Group criteria. All efficacy data will be confirmed by an Independent Adjudication Committee. Results: Between January 2009 and October 2010, 143 patients were enrolled from 41 centers in 12 countries across Asia-Pacific, Europe, and North America. Median duration of MM for the entire study population was 4.6 years, including 31% whose disease had progressed on or within 60 days of last therapy and 66% whose disease had achieved < 25% response to the regimen immediately preceding study entry. The study population was heavily pretreated, having received a median of 4 prior lines of therapy (range 2–17, ≥ 4 prior regimens: 69%). Prior anti-myeloma agents included 100% BTZ (median 2 prior regimens containing BTZ), 100% IMiDs (thalidomide [85%], lenalidomide [71%], or pomalidomide [4%]), and 74% stem cell transplant. As of July 2011, 142 of the enrolled patients received study medication, with a median exposure of 4 cycles (mean 6.2 cycles; range 1–26 cycles). The most common treatment-emergent adverse events regardless of relationship to study drug were predominantly hematologic and gastrointestinal disorders. Of interest, peripheral neuropathy was infrequent and grade ≥ 3 PN occurred in 2 patients (1.6%). The final efficacy evaluation of the IAC will occur in September 2011. Conclusions: Final data for all primary and secondary endpoints, including response assessment and time-to-event data for PFS/TTP, OS, and duration of response will be available at the annual ASH meeting. Disclosures: Siegel: Millenium: Honoraria, Research Funding, Speakers Bureau; Merck: Honoraria. Off Label Use: Vorinostat, an inhibitor of histone deacetylase, is approved in the US for the treatment of cutaneous manifestations in patients with cutaneous T cell lymphoma (CTCL) who have progressive, persistent or recurrent disease on or following two systemic therapies. Vorinostat is currently under investigation for the treatment of relapsed malignant pleural mesothelioma, relapsed/refractory B cell lymphoma (in combination with other chemotherapy agents), and relapsed/refractory multiple myeloma (in combination with bortezomib and other chemotherapy agents). Dimopoulos:Celgene, Ortho-Biotech: Consultancy, Honoraria. Yoon:NK Bio: Consultancy; Celgene: Consultancy. Kaufman:Merck; Celgene: Research Funding; Millenium; Onxy; Novartis; Keryx: Consultancy. Goldschmidt:Amgen, Novartis, Chugai: Research Funding; Janssen-Cilag, Celgene: Membership on an entity's Board of Directors or advisory committees, Research Funding. Reece:Merck: Honoraria, Research Funding; Janssen: Honoraria, Research Funding; Celgene: Honoraria, Research Funding; Otsuka: Honoraria, Research Funding. Leleu:Janssen Cilag, Celgene, Novartis-Amgen, Leo Pharma, Chugai, Roche: Honoraria, Research Funding. Cavo:Millennium: Honoraria, Membership on an entity's Board of Directors or advisory committees; Celgene: Honoraria, Membership on an entity's Board of Directors or advisory committees; Janssen: Honoraria, Membership on an entity's Board of Directors or advisory committees, Speakers Bureau. Jagannath:Merck: Membership on an entity's Board of Directors or advisory committees; Celgene: Membership on an entity's Board of Directors or advisory committees; Millennium: Membership on an entity's Board of Directors or advisory committees. Graef:Merck: Employment. Houp:Merck: Employment. Sun:Merck & Co., Inc.: Employment. Howe:Merck: Employment. Anderson:Celgene: Consultancy; Millennium: Consultancy; Novartis: Consultancy; BMS: Consultancy; Onyx: Consultancy; Merck: Consultancy; Acetylon: .


Blood ◽  
2018 ◽  
Vol 132 (Supplement 1) ◽  
pp. 4973-4973
Author(s):  
Gordon Haire ◽  
Karl Egan ◽  
Barry Kevane ◽  
Michael Fay ◽  
Anne Fortune ◽  
...  

Abstract Background Eisenmenger Syndrome (ES) is an extremely rare and serious complication of congenital heart disease in which pulmonary vascular resistance overcomes that of the systemic circulation causing bidirectional or reversed flow across an intracardiac shunt. Patients with ES have an increased incidence of both thrombotic and bleeding complications which occur through poorly understood mechanisms. We have recently demonstrated a critical mechanistic role for platelets in supporting abnormal hypercoagulability in patients with ES using calibrated automated thrombography (Kevane & Ní Áinle et al., J Thromb Haemost 2018). Aims Based upon our published data, we further hypothesized that patients with ES would have higher plasma concentrations of platelet activation markers. Therefore, the aim of this study was to measure plamsa levels of soluble P-Selectin (sP-Selectin) and soluble glycoprotein VI (sGPVI) in patients with ES and in matched healthy controls. Methods Patients over the age of 18 with Eisenmenger Syndrome and healthy controls were recruited from the cardiology service at a large tertiary referral centre (Mater Misericordiae University Hospital) incorporating the Irish national congenital heart disease service. Platelet poor plasma (PPP) was generated from participants' blood by centrifugation at 2000xg for 10 minutes at room temperature. Commercially available ELISA assays employing quantitative sandwich immunoassay techniques were performed according to manufacturer protocol to measure plasma sP-Selectin and sGPVI levels. All experiments were performed in duplicate with results expressed as mean +/- standard error of mean. Comparisons between groups were made utilising the Student's t-test with a p-value < 0.05 deemed to represent statistical significance. Results During the study period, >14,000 patients attended the MMUH cardiology service. Of these, 14 consecutive patients with a diagnosis of ES and 10 matched healthy controls were recruited. Patients with ES had significantly elevated plasma concentrations of both soluble P-Selectin [38.5 +/- 8.3 vs. 11.3 +/- 4.2 ng/ml (p < 0.05)] and soluble GPVI [10.5 +/- 2.4 vs. 1.5 +/- 1.4 ng/ml (p < 0.01)] compared to healthy controls. Conclusion Building upon our recently published data in this population, a large cohort given the extreme rarity of this condition, increased concentrations of markers of platelet activation in individuals with ES further suggest a mechanistic role for platelets in ES-mediated hypercoagulability. This phenotype may be a consequence of vascular pathology associated with pulmonary hypertension in these patients. As such, therapies targeted at underlying vascular pathologies may act to ameliorate prothrombotic tendencies and we aim to characterize response to therapy during this ongoing study. Disclosures Ni Ainle: Leo Pharma: Research Funding; Actelion: Research Funding; Bayer: Research Funding; Bayer: Membership on an entity's Board of Directors or advisory committees; BMS: Membership on an entity's Board of Directors or advisory committees; Daiichi Sankyo: Membership on an entity's Board of Directors or advisory committees; Boehringer: Membership on an entity's Board of Directors or advisory committees.


Blood ◽  
2017 ◽  
Vol 130 (Suppl_1) ◽  
pp. 808-808
Author(s):  
Julia Wells ◽  
Tianyu Cai ◽  
Cécile Schiffer-Manniou ◽  
Stéphanie Filipe ◽  
Agnès Gouble ◽  
...  

Abstract Autologous T-cells engineered with chimeric antigen receptors (CARs) against CD19 are proving to be an efficacious immunotherapy for patients with relapsed or refractory B-cell acute lymphoblastic leukemia (B-ALL). At present, CAR technology is administered through the custom-made manufacturing of therapeutic products from each patient's own T-cells. However, this patient-specific autologous paradigm is a significant limiting factor in the large-scale deployment of CAR technology. In this study, we utilized allogeneic "off-the-shelf" engineered CAR T-cells from third-party healthy donors. The CD22 surface antigen is commonly expressed in B-ALL patients as well as in healthy B-cells. Here, its potential as a CAR target was investigated using allogeneic off-the shelf engineered CAR T-cells against human CD22 (UCART22). UCART22 cells harbor surface expression of an anti-CD22 CAR (CD22 scFv-41BB-CD3z) and the RQR8 ligand, a safety feature rendering the T-cells sensitive to the monoclonal antibody rituximab. To reduce the potential for alloreactivity, the cell surface expression of the T-cell receptor (TCR) is abrogated through the inactivation of the TCRα constant (TRAC) gene using Cellectis' TALEN® gene-editing technology. The level of CD22 cell surface molecules was measured using BD Quantbrite beads for both patient peripheral blood samples and B-ALL cell lines. B-ALL cell lines (n=8) expressed a greater amount of CD22 molecules per cell than patient samples (n=14) (5,028 +/- 1,342 compared to 951 +/-160 molecules/cell, p=0.044), with highest expression of CD22 in two Ph-like B-ALL cell lines (MUTZ5, shown in Figure1A and MHH-CALL4). The in vitro cytotoxic activity of UCART22 cells was evaluated by co-culturing UCART22 or non-transduced CAR(-) TCRαβ(-) control T-cells (NTD) with B-ALL cell lines and primary human samples, at a maximum 10:1 effector to target ratio (represented in Figure1B). Using flow cytometry, significant antigen-specific cytotoxic activity of UCART22 cells was found compared to NTD controls and correlated with CD22 expression factored by the %kolmogorov-smirnov max difference in CD22-PE fluorescence compared to unstained controls (Pearson correlation r-squared for cell lines= 0.6850, p=0.0001 and r-squared for patient samples=0.6204, p=0.0008). Secretion of 13 cytokines was measured after 1:1 co-incubation of effector and target cells. UCART22 cells stimulated by CD22(+) B-ALL, but not NTD cells, secreted high levels of IFNγ, TNFα, IL-5, IL-17A and IL-17F in the culture supernatants, with cytokine levels being proportionate to CD22 abundance (represented in Figure1C). In addition, immune compromised mice engrafted with Daudi cells, a CD22(+) expressing Burkitt's lymphoma cell line, were treated with UCART22 cells. Treatment doses of 1-10x10^6 cells per mouse reduced disease burden (Figure 1D), measured by bioluminescence imaging, and extended survival in a dose-dependent fashion compared to saline or NTD treated controls. Additional PDX studies using B-ALL patient derived xenografts are ongoing and will be presented. Altogether, these results show supporting evidence for the future use of allogenic UCART22 in B-ALL immunotherapy. Disclosures Schiffer-Manniou: Cellectis SA: Employment. Filipe: Cellectis: Employment. Gouble: Cellectis SA: Employment. Galetto: Cellectis SA: Employment. Jain: ADC Therapeutics: Honoraria, Membership on an entity's Board of Directors or advisory committees, Research Funding; Novartis: Honoraria, Membership on an entity's Board of Directors or advisory committees; Celgene: Research Funding; Verastem: Research Funding; Pfizer: Honoraria, Membership on an entity's Board of Directors or advisory committees, Research Funding; Pharmacyclics: Honoraria, Membership on an entity's Board of Directors or advisory committees, Research Funding; BMS: Research Funding; Abbvie: Research Funding; Incyte: Research Funding; Genentech: Research Funding; Novimmune: Honoraria, Membership on an entity's Board of Directors or advisory committees; Servier: Honoraria, Membership on an entity's Board of Directors or advisory committees, Research Funding; Adaptive Biotechnologies: Honoraria, Membership on an entity's Board of Directors or advisory committees. Jabbour: Bristol-Myers Squibb: Consultancy. Smith: Cellectis Inc: Employment.


Blood ◽  
2015 ◽  
Vol 126 (23) ◽  
pp. 1114-1114
Author(s):  
Karl Egan ◽  
Eimear Dunne ◽  
Audrey Dillon ◽  
Barry Kevane ◽  
Zita Galvin ◽  
...  

Abstract Background Cirrhosis is a consequence of prolonged inflammation arising from chronic liver disease of different aetiologies. It is characterised by tissue fibrosis, the deposition of collagen-rich extracellular matrix tissue within the liver. Glycoprotein VI (GPVI) is platelet-specific collagen receptor that is shed from the platelet surface in a metalloproteinase-dependent manner in response to GPVI ligation. The shed extracellular region of GPVI can be detected in plasma and used as a measure of GPVI-dependent platelet activation in vivo. Several lines of evidence suggest that GPVI-dependent platelet activation occurs in cirrhosis. Platelets have been shown to accumulate at sites of collagen-rich fibrotic tissue. Circulating levels of collagen are increased in cirrhosis. Collagen-induced platelet aggregation responses are reduced in vitro with cirrhosis. Based on these results, we hypothesised that soluble GPVI (sGPVI) levels are increased in patients with cirrhosis. As such, the aim of this study was to quantify sGPVI levels in patients with cirrhosis and compare to healthy controls. Methods Compensated cirrhotic patients were recruited at the Mater Misericordiae University Hospital, Dublin, Ireland. The diagnosis of cirrhosis was based on clinical examination, blood tests, and radiological examination (nodular surface, larger right lobe, coarse echotexture). Exclusion criteria were decompensated cirrhosis, recent thrombotic events, and antiplatelet and/or anticoagulant therapies. Healthy controls were recruited at the Royal College of Surgeons in Ireland and Beaumont Hospital, Dublin, Ireland. Blood samples were collected into vacutainers containing 3.2 % sodium citrate as anticoagulant. sGPVI levels in platelet poor plasma were measured using an in house custom ELISA. Results 57 patients with mixed aetiology cirrhosis and 55 healthy controls were recruited. In the patient group, 42% of patients had alcoholic liver disease (ALD), 30% had hepatitis C (HCV), 7% had non alcoholic fatty liver disease (NAFLD), 5% had Hepatitis B (HBV), 5% had autoimmune hepatitis (AIH), 5% had cryptogenic liver disease, 4% had hereditary haemochromatosis (HH), and 2% had primary biliary cholangitis (PBC). sGPVI levels were significantly increased in patients with cirrhosis (5.8 ± 0.6 ng/ml, n = 57) compared to healthy controls (3.2 ± 0.4 ng/ml, n = 55, p < 0.0001). There was no significant difference between sGPVI levels in AIH (4 ± 1 ng/ml, n = 3), ALD (5.6 ± 1 ng/ml, n = 24), cryptogenic (12 ± 5 ng/ml, n = 3), HBV (3.1 ± 1 ng/ml, n = 3), HCV (5 ± 0.6 ng/ml), or NAFLD (5.3 ± 1.1 ng/ml, n = 4). sGPVI levels did not correlate with platelet count (r = 0.12, p = 0.3) or parameters of liver cell function (albumin, bilirubin, prothrombin time, and liver stiffness measurements). Conclusion sGPVI levels are significantly increased in patients with mixed aetiology cirrhosis. This indicates collagen induced platelet activation is occurring in vivo and suggests the presence of an underlying coagulopathy in patients with cirrhosis. Disclosures Ní Áinle: Actelion Pharma: Research Funding; Leo Pharma: Research Funding; Bristol Myers Squibb: Membership on an entity's Board of Directors or advisory committees; Bayer: Membership on an entity's Board of Directors or advisory committees; Daiichi Sankyo: Membership on an entity's Board of Directors or advisory committees; Boehringer Ingelheim: Membership on an entity's Board of Directors or advisory committees.


Sign in / Sign up

Export Citation Format

Share Document